Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters

Therapeutic Methods and Therapies TCIM
Language
Affiliation country
Publication year range
1.
Altern Ther Health Med ; 29(3): 16-25, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36580671

ABSTRACT

Aim: The aim of this study was to elucidate the potential mechanism of Rg1 in alleviating hepatic ischemia-reperfusion (HIRI) through the mitophagy pathway. Methods: The HIRI rat models were established and divided into 4 groups: the sham group, sham+Rg1 group, ischemia/perfusion (I/R) group and I/R+Rg1 group. Then the activities of aspartate transaminase (AST) and alanine aminotransferase (ALT) were detected by automatic serum analyzer. Meanwhile, cell apoptosis and changes in liver tissues were checked by TUNEL assay and histopathological analysis, respectively. The relative protein levels were detected by western blotting. Subsequently, cell counting Kit-8 assay and cytometric analysis were used to investigate cell viability and apoptosis of liver cells. Finally, the time points of the strongest mitochondrial autophagy were explored and the mitochondrial morphology was observed by the mitochondrial transmembrane potential (MMP) in vivo and in vitro. Results: The mitophagy aggravated hepatocyte damage during liver I/R in vivo. In addition, Rg1 alleviated liver damage after liver I/R, maintained the stability of MMP and inhibited mitochondrial autophagy and signaling pathways during liver I/R in vivo. Furthermore, Rg1 could effectively increase cell viability, inhibit cell apoptosis and stabilize MMP after OGD/R injury in vitro Moreover, Rg1 exerted its protective effect on HIRI by regulating the PINK1/Parkin signaling pathway and the mitochondrial autophagy. Conclusion: Rg1 could further improve its mechanism of alleviating HIRI in apoptosis and autophagy, 2 types of regulated programmed cell death via the mitochondrial pathway.


Subject(s)
Reperfusion Injury , Rats , Animals , Rats, Sprague-Dawley , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Ischemia/metabolism , Ischemia/pathology , Liver/metabolism , Apoptosis , Autophagy , Reperfusion
2.
Am J Chin Med ; 48(5): 1159-1178, 2020.
Article in English | MEDLINE | ID: mdl-32668973

ABSTRACT

Hepatic ischemia-reperfusion (IR) injury remains the major cause of liver damage post-liver surgery or transplantation. Diminishing oxidative stress and inflammatory responses is a powerful channel to reduce the rate of morbidity and mortality. Gastrodin (GSTD), a bioactive compound extracted from the traditional Chinese herbal agent with a long history of clinical application in nervous system diseases, is suggested to possess anti-oxidative effects on liver diseases, such as nonalcoholic fatty liver disease. However, the therapeutic potential of GSTD in liver IR injury remains unclear. In this paper, we performed surgery to set up the 70% hepatic IR injury models in mice after a three-day pretreatment of GSTD. We found the administration of GSTD reduced liver damage, which correlated with lower histological Suzuki's score, lower serum alanine transaminase (AST) and alanine transaminase (ALT) levels, less oxidative stress, and cell apoptosis in a dose-responsive manner, as compared to the parallel control. Meanwhile, we observed a great induction of heme oxygenase-1 (HO-1) and an activation of the p38 mitogen-activated protein kinases/nuclear factor erythroid 2-related factor 2 (p38MAPK/Nrf2) pathway in response to the GSTD pretreatment, while the protective effects upon GSTD diminished in mice with HO-1 heterozygous mutation. In addition, GSTD inhibited IR induced toll-like receptor (TLR) 4, but not TLR2 in a HO-1 dependent manner, leading to a down-regulation of cytokines, such as interleukin (IL)-6 and TNF-[Formula: see text]. Collectively, our findings revealed GSTD attenuated liver IR injury via activation of the HO-1 pathway, providing a novel therapeutic strategy to minimize the IR induced oxidative stress in the process of liver transplantation.


Subject(s)
Antioxidants , Benzyl Alcohols/administration & dosage , Benzyl Alcohols/pharmacology , Glucosides/administration & dosage , Glucosides/pharmacology , Liver , NF-E2-Related Factor 2/metabolism , Phytotherapy , Reperfusion Injury/prevention & control , Signal Transduction/drug effects , Animals , Cytokines/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Drugs, Chinese Herbal , Heme Oxygenase-1/metabolism , Mice, Inbred C57BL , Oxidative Stress/drug effects , Preoperative Care , Toll-Like Receptor 4/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Braz J Med Biol Res ; 51(10): e7439, 2018 Aug 16.
Article in English | MEDLINE | ID: mdl-30156611

ABSTRACT

Nuclear factor erythroid-related factor 2 (Nrf2) has been implicated in several detoxifying and antioxidant defense processes. Nrf2-mediated heme oxygenase-1 (HO-1) expression was demonstrated to play a key role against oxidative stress. Gastrodin (GSTD) is a well-known active compound isolated from the roots of Rhizoma gastrodiae, a plant used in ancient Chinese traditional medicine. The aim of this work was to investigate whether GSTD could alleviate H2O2-induced oxidative stress in mouse liver sinusoidal endothelial cells (LSECs). In LSECs exposed to 1 mM H2O2, treatment with GSTD (1, 10, or 50 µM) resulted in higher cell viability than the untreated control. Treated cells maintained a higher Bcl2/Bax ratio and suppressed caspase-9 expression compared with untreated cells, reducing cell apoptosis. GSTD was protective for H2O2-induced oxidative injury by reducing the generation of intracellular reactive oxygen species and malondialdehyde. HO-1 and Nrf2 expressions were synergistically upregulated by GSTD. Inhibition of HO-1 by 10 µM zinc protoporphyrin resulted in less protective effects on cell viability and malondialdehyde reduction by GSTD treatment in H2O2-exposed LSECs. Additionally, phosphorylated p38 in LSECs exposed to H2O2 was elevated by GSTD. Inhibition of p38 phosphorylation by SB203580 did not induce Nrf2 and HO-1 expression after 1 or 10 µM GSTD treatment and the protective effect on cell viability and malondialdehyde reduction in H2O2-exposed LSECs was reduced. The data conclusively demonstrated that GSTD-induced HO-1 and Nrf2 expression is involved in protection of LSECs from H2O2-induced oxidative injury, which may be regulated by p38 phosphorylation.


Subject(s)
Benzyl Alcohols/pharmacology , Endothelial Cells/drug effects , Glucosides/pharmacology , Heme Oxygenase-1/metabolism , Hydrogen Peroxide/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Liver/cytology , Liver/drug effects , Malondialdehyde/metabolism , Mice , Models, Theoretical , Oxidative Stress/physiology , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects , p38 Mitogen-Activated Protein Kinases/blood
4.
Braz. j. med. biol. res ; 51(10): e7439, 2018. graf
Article in English | LILACS | ID: biblio-951707

ABSTRACT

Nuclear factor erythroid-related factor 2 (Nrf2) has been implicated in several detoxifying and antioxidant defense processes. Nrf2-mediated heme oxygenase-1 (HO-1) expression was demonstrated to play a key role against oxidative stress. Gastrodin (GSTD) is a well-known active compound isolated from the roots of Rhizoma gastrodiae, a plant used in ancient Chinese traditional medicine. The aim of this work was to investigate whether GSTD could alleviate H2O2-induced oxidative stress in mouse liver sinusoidal endothelial cells (LSECs). In LSECs exposed to 1 mM H2O2, treatment with GSTD (1, 10, or 50 µM) resulted in higher cell viability than the untreated control. Treated cells maintained a higher Bcl2/Bax ratio and suppressed caspase-9 expression compared with untreated cells, reducing cell apoptosis. GSTD was protective for H2O2-induced oxidative injury by reducing the generation of intracellular reactive oxygen species and malondialdehyde. HO-1 and Nrf2 expressions were synergistically upregulated by GSTD. Inhibition of HO-1 by 10 µM zinc protoporphyrin resulted in less protective effects on cell viability and malondialdehyde reduction by GSTD treatment in H2O2-exposed LSECs. Additionally, phosphorylated p38 in LSECs exposed to H2O2 was elevated by GSTD. Inhibition of p38 phosphorylation by SB203580 did not induce Nrf2 and HO-1 expression after 1 or 10 µM GSTD treatment and the protective effect on cell viability and malondialdehyde reduction in H2O2-exposed LSECs was reduced. The data conclusively demonstrated that GSTD-induced HO-1 and Nrf2 expression is involved in protection of LSECs from H2O2-induced oxidative injury, which may be regulated by p38 phosphorylation.


Subject(s)
Animals , Rabbits , Benzyl Alcohols/pharmacology , Endothelial Cells/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Heme Oxygenase-1/metabolism , Glucosides/pharmacology , Hydrogen Peroxide/pharmacology , Up-Regulation/drug effects , Cell Survival/drug effects , Apoptosis/drug effects , Liver/cytology , Liver/drug effects , Malondialdehyde/metabolism , Models, Theoretical
SELECTION OF CITATIONS
SEARCH DETAIL